Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(27): e2402259121, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38917012

RESUMO

HCN1-4 channels are the molecular determinants of the If/Ih current that crucially regulates cardiac and neuronal cell excitability. HCN dysfunctions lead to sinoatrial block (HCN4), epilepsy (HCN1), and chronic pain (HCN2), widespread medical conditions awaiting subtype-specific treatments. Here, we address the problem by solving the cryo-EM structure of HCN4 in complex with ivabradine, to date the only HCN-specific drug on the market. Our data show ivabradine bound inside the open pore at 3 Å resolution. The structure unambiguously proves that Y507 and I511 on S6 are the molecular determinants of ivabradine binding to the inner cavity, while F510, pointing outside the pore, indirectly contributes to the block by controlling Y507. Cysteine 479, unique to the HCN selectivity filter (SF), accelerates the kinetics of block. Molecular dynamics simulations further reveal that ivabradine blocks the permeating ion inside the SF by electrostatic repulsion, a mechanism previously proposed for quaternary ammonium ions.


Assuntos
Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Ivabradina , Simulação de Dinâmica Molecular , Ivabradina/química , Ivabradina/farmacologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/química , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/antagonistas & inibidores , Humanos , Microscopia Crioeletrônica , Animais , Canais de Potássio/química , Canais de Potássio/metabolismo , Proteínas Musculares/química , Proteínas Musculares/metabolismo
2.
Elife ; 112022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35315774

RESUMO

Tongmai Yangxin (TMYX) is a complex compound of the Traditional Chinese Medicine (TCM) used to treat several cardiac rhythm disorders; however, no information regarding its mechanism of action is available. In this study we provide a detailed characterization of the effects of TMYX on the electrical activity of pacemaker cells and unravel its mechanism of action. Single-cell electrophysiology revealed that TMYX elicits a reversible and dose-dependent (2/6 mg/ml) slowing of spontaneous action potentials rate (-20.8/-50.2%) by a selective reduction of the diastolic phase (-50.1/-76.0%). This action is mediated by a negative shift of the If activation curve (-6.7/-11.9 mV) and is caused by a reduction of the cyclic adenosine monophosphate (cAMP)-induced stimulation of pacemaker channels. We provide evidence that TMYX acts by directly antagonizing the cAMP-induced allosteric modulation of the pacemaker channels. Noticeably, this mechanism functionally resembles the pharmacological actions of muscarinic stimulation or ß-blockers, but it does not require generalized changes in cytoplasmic cAMP levels thus ensuring a selective action on rate. In agreement with a competitive inhibition mechanism, TMYX exerts its maximal antagonistic action at submaximal cAMP concentrations and then progressively becomes less effective thus ensuring a full contribution of If to pacemaker rate during high metabolic demand and sympathetic stimulation.


Assuntos
AMP Cíclico , Sistemas do Segundo Mensageiro , Potenciais de Ação , Animais , China , AMP Cíclico/metabolismo , Miócitos Cardíacos/metabolismo , Coelhos
3.
Mol Cell ; 81(14): 2929-2943.e6, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34166608

RESUMO

The HCN1-4 channel family is responsible for the hyperpolarization-activated cation current If/Ih that controls automaticity in cardiac and neuronal pacemaker cells. We present cryoelectron microscopy (cryo-EM) structures of HCN4 in the presence or absence of bound cAMP, displaying the pore domain in closed and open conformations. Analysis of cAMP-bound and -unbound structures sheds light on how ligand-induced transitions in the channel cytosolic portion mediate the effect of cAMP on channel gating and highlights the regulatory role of a Mg2+ coordination site formed between the C-linker and the S4-S5 linker. Comparison of open/closed pore states shows that the cytosolic gate opens through concerted movements of the S5 and S6 transmembrane helices. Furthermore, in combination with molecular dynamics analyses, the open pore structures provide insights into the mechanisms of K+/Na+ permeation. Our results contribute mechanistic understanding on HCN channel gating, cyclic nucleotide-dependent modulation, and ion permeation.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ativação do Canal Iônico/fisiologia , Íons/metabolismo , Proteínas Musculares/metabolismo , Canais de Potássio/metabolismo , Linhagem Celular , Microscopia Crioeletrônica/métodos , AMP Cíclico/metabolismo , Células HEK293 , Humanos
4.
Elife ; 72018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29923826

RESUMO

Binding of TRIP8b to the cyclic nucleotide binding domain (CNBD) of mammalian hyperpolarization-activated cyclic nucleotide-gated (HCN) channels prevents their regulation by cAMP. Since TRIP8b is expressed exclusively in the brain, we envisage that it can be used for orthogonal control of HCN channels beyond the central nervous system. To this end, we have identified by rational design a 40-aa long peptide (TRIP8bnano) that recapitulates affinity and gating effects of TRIP8b in HCN isoforms (hHCN1, mHCN2, rbHCN4) and in the cardiac current If in rabbit and mouse sinoatrial node cardiomyocytes. Guided by an NMR-derived structural model that identifies the key molecular interactions between TRIP8bnano and the HCN CNBD, we further designed a cell-penetrating peptide (TAT-TRIP8bnano) which successfully prevented ß-adrenergic activation of mouse If leaving the stimulation of the L-type calcium current (ICaL) unaffected. TRIP8bnano represents a novel approach to selectively control HCN activation, which yields the promise of a more targeted pharmacology compared to pore blockers.


Assuntos
AMP Cíclico/química , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/química , Miócitos Cardíacos/efeitos dos fármacos , Peptídeos/farmacologia , Canais de Potássio/química , Animais , Sítios de Ligação , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/metabolismo , AMP Cíclico/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Peptídeos/síntese química , Peroxinas/química , Peroxinas/genética , Peroxinas/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Coelhos , Nó Sinoatrial/citologia , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
5.
Transl Res ; 192: 54-67, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29245016

RESUMO

Atrial fibrillation (AF) is characterized by electrical, contractile, and structural remodeling mediated by interstitial fibrosis. It has been shown that human cardiac mesenchymal progenitor cells (CMPCs) can be differentiated into endothelial, smooth muscle, and fibroblast cells. Here, we have investigated, for the first time, the contribution of CMPCs in the fibrotic process occurring in AF. As expected, right auricolae samples displayed significantly higher fibrosis in AF vs control (CTR) patients. In tissue samples of AF patients only, double staining for c-kit and the myofibroblast marker α-smooth muscle actin (α-SMA) was detected. The number of c-kit-positive CMPC was higher in atrial subepicardial regions of CTR than AF cells. AF-derived CMPC (AF-CMPC) and CTR-derived CMPC (Ctr-CMPC) were phenotypically similar, except for CD90 and c-kit, which were significantly more present in AF and CTR cells, respectively. Moreover, AF showed a lower rate of population doubling and fold enrichment vs Ctr-CMPC. When exogenously challenged with the profibrotic transforming growth factor-ß1 (TGF-ß1), AF-CMPC showed a significantly higher nuclear translocation of SMAD2 than Ctr-CMPC. In addition, TGF-ß1 treatment induced the upregulation of COL1A1 and COL1A2 in AF-CMPC only. Further, both a marked production of soluble collagen and α-SMA upregulation have been observed in AF-CMPC only. Finally, electrophysiological studies showed that the inwardly rectifying potassium current (IK1) was evenly present in AF- and Ctr-CMPC in basal conditions and similarly disappeared after TGF-ß1 exposure. All together, these data suggest that AF steers the resident atrial CMPC compartment toward an electrically inert profibrotic phenotype.


Assuntos
Fibrilação Atrial/patologia , Células-Tronco Mesenquimais/patologia , Miocárdio/patologia , Miofibroblastos/patologia , Idoso , Fibrilação Atrial/fisiopatologia , Diferenciação Celular , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Fator de Crescimento Transformador beta1/farmacologia
7.
J Interv Card Electrophysiol ; 46(1): 19-28, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26781742

RESUMO

BACKGROUND: Together with the afferent branches of the autonomic nervous system, the sinoatrial node (SAN) forms a functional unit whose function is to fire rhythmic action potentials at a rate optimal for coping with the metabolic needs of the body. Dysfunctional behavior of this complex unit may thus result in SAN rhythm disorders. Among these disorders, there is the inappropriate sinus tachycardia (IST) which occurs when an unjustified fast SAN rate is present. METHODS: We here present a critical review of the role of pacemaker f/HCN channels in cardiac rhythm generation and modulation and their involvement in IST. RESULTS: Recent evidence demonstrates that a familial form of IST is associated with a gain-of-function mutation in the HCN4 pacemaker channel (R524Q) which confers an increased sensitivity to the second messenger cAMP, a key mediator in sympathetic modulation. CONCLUSIONS: This finding is consistent with the general view that hypersympathetic tone is one of the causes of IST and introduces the novel concept of defective funny channel-dependent tachyarrhythmias.


Assuntos
Potenciais de Ação , Relógios Biológicos , Frequência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Nó Sinoatrial/fisiopatologia , Taquicardia Sinusal/fisiopatologia , Animais , AMP Cíclico/metabolismo , Humanos , Ativação do Canal Iônico , Modelos Cardiovasculares
8.
Nat Chem Biol ; 10(6): 457-62, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24776929

RESUMO

cAMP mediates autonomic regulation of heart rate by means of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which underlie the pacemaker current If. cAMP binding to the C-terminal cyclic nucleotide binding domain enhances HCN open probability through a conformational change that reaches the pore via the C-linker. Using structural and functional analysis, we identified a binding pocket in the C-linker of HCN4. Cyclic dinucleotides, an emerging class of second messengers in mammals, bind the C-linker pocket (CLP) and antagonize cAMP regulation of the channel. Accordingly, cyclic dinucleotides prevent cAMP regulation of If in sinoatrial node myocytes, reducing heart rate by 30%. Occupancy of the CLP hence constitutes an efficient mechanism to hinder ß-adrenergic stimulation on If. Our results highlight the regulative role of the C-linker and identify a potential drug target in HCN4. Furthermore, these data extend the signaling scope of cyclic dinucleotides in mammals beyond their first reported role in innate immune system.


Assuntos
AMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Fosfatos de Dinucleosídeos/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ativação do Canal Iônico/fisiologia , Proteínas Musculares/metabolismo , Canais de Potássio/metabolismo , Animais , Sítios de Ligação , Western Blotting , Cristalografia por Raios X , GMP Cíclico/química , GMP Cíclico/metabolismo , Fosfatos de Dinucleosídeos/química , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Estrutura Molecular , Proteínas Musculares/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio/genética , Nó Sinoatrial/citologia , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Transfecção
9.
Circ Res ; 113(4): 389-98, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23753573

RESUMO

RATIONALE: A cell-based biological pacemaker is based on the differentiation of stem cells and the selection of a population displaying the molecular and functional properties of native sinoatrial node (SAN) cardiomyocytes. So far, such selection has been hampered by the lack of proper markers. CD166 is specifically but transiently expressed in the mouse heart tube and sinus venosus, the prospective SAN. OBJECTIVE: We have explored the possibility of using CD166 expression for isolating SAN progenitors from differentiating embryonic stem cells. METHODS AND RESULTS: We found that in embryonic day 10.5 mouse hearts, CD166 and HCN4, markers of the pacemaker tissue, are coexpressed. Sorting embryonic stem cells for CD166 expression at differentiation day 8 selects a population of pacemaker precursors. CD166+ cells express high levels of genes involved in SAN development (Tbx18, Tbx3, Isl-1, Shox2) and function (Cx30.2, HCN4, HCN1, CaV1.3) and low levels of ventricular genes (Cx43, Kv4.2, HCN2, Nkx2.5). In culture, CD166+ cells form an autorhythmic syncytium composed of cells morphologically similar to and with the electrophysiological properties of murine SAN myocytes. Isoproterenol increases (+57%) and acetylcholine decreases (-23%) the beating rate of CD166-selected cells, which express the ß-adrenergic and muscarinic receptors. In cocultures, CD166-selected cells are able to pace neonatal ventricular myocytes at a rate faster than their own. Furthermore, CD166+ cells have lost pluripotency genes and do not form teratomas in vivo. CONCLUSIONS: We demonstrated for the first time the isolation of a nonteratogenic population of cardiac precursors able to mature and form a fully functional SAN-like tissue.


Assuntos
Molécula de Adesão de Leucócito Ativado/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Miócitos Cardíacos/citologia , Nó Sinoatrial/citologia , Células-Tronco/citologia , Acetilcolina/farmacologia , Animais , Biomarcadores/metabolismo , Cardiotônicos/farmacologia , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Técnicas de Cocultura , Células-Tronco Embrionárias/efeitos dos fármacos , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Isoproterenol/farmacologia , Camundongos , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
10.
PLoS One ; 7(12): e51694, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284745

RESUMO

Adult human cardiac mesenchymal-like stromal cells (CStC) represent a relatively accessible cell type useful for therapy. In this light, their conversion into cardiovascular precursors represents a potential successful strategy for cardiac repair. The aim of the present work was to reprogram CStC into functionally competent cardiovascular precursors using epigenetically active small molecules. CStC were exposed to low serum (5% FBS) in the presence of 5 µM all-trans Retinoic Acid (ATRA), 5 µM Phenyl Butyrate (PB), and 200 µM diethylenetriamine/nitric oxide (DETA/NO), to create a novel epigenetically active cocktail (EpiC). Upon treatment the expression of markers typical of cardiac resident stem cells such as c-Kit and MDR-1 were up-regulated, together with the expression of a number of cardiovascular-associated genes including KDR, GATA6, Nkx2.5, GATA4, HCN4, NaV1.5, and α-MHC. In addition, profiling analysis revealed that a significant number of microRNA involved in cardiomyocyte biology and cell differentiation/proliferation, including miR 133a, 210 and 34a, were up-regulated. Remarkably, almost 45% of EpiC-treated cells exhibited a TTX-sensitive sodium current and, to a lower extent in a few cells, also the pacemaker I(f) current. Mechanistically, the exposure to EpiC treatment introduced global histone modifications, characterized by increased levels of H3K4Me3 and H4K16Ac, as well as reduced H4K20Me3 and H3s10P, a pattern compatible with reduced proliferation and chromatin relaxation. Consistently, ChIP experiments performed with H3K4me3 or H3s10P histone modifications revealed the presence of a specific EpiC-dependent pattern in c-Kit, MDR-1, and Nkx2.5 promoter regions, possibly contributing to their modified expression. Taken together, these data indicate that CStC may be epigenetically reprogrammed to acquire molecular and biological properties associated with competent cardiovascular precursors.


Assuntos
Diferenciação Celular , Epigênese Genética/genética , Coração/fisiologia , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Adulto , Biomarcadores/metabolismo , Western Blotting , Cromatina/genética , Imunoprecipitação da Cromatina , Eletrofisiologia , Perfilação da Expressão Gênica , Histonas/metabolismo , Humanos , Técnicas In Vitro , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Doadores de Óxido Nítrico/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Tretinoína/farmacologia , Regulação para Cima
11.
Am J Physiol Heart Circ Physiol ; 300(5): H1875-84, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21357510

RESUMO

The efficacy of cardiac repair by stem cell administration relies on a successful functional integration of injected cells into the host myocardium. Safety concerns have been raised about the possibility that stem cells may induce foci of arrhythmia in the ischemic myocardium. In a previous work (36), we showed that human cord blood CD34(+) cells, when cocultured on neonatal mouse cardiomyocytes, exhibit excitation-contraction coupling features similar to those of cardiomyocytes, even though no human genes were upregulated. The aims of the present work are to investigate whether human CD34(+) cells, isolated after 1 wk of coculture with neonatal ventricular myocytes, possess molecular and functional properties of cardiomyocytes and to discriminate, using a reporter gene system, whether cardiac differentiation derives from a (trans)differentiation or a cell fusion process. Umbilical cord blood CD34(+) cells were isolated by a magnetic cell sorting method, transduced with a lentiviral vector carrying the enhanced green fluorescent protein (EGFP) gene, and seeded onto primary cultures of spontaneously beating rat neonatal cardiomyocytes. Cocultured EGFP(+)/CD34(+)-derived cells were analyzed for their electrophysiological features at different time points. After 1 wk in coculture, EGFP(+) cells, in contact with cardiomyocytes, were spontaneously contracting and had a maximum diastolic potential (MDP) of -53.1 mV, while those that remained isolated from the surrounding myocytes did not contract and had a depolarized resting potential of -11.4 mV. Cells were then resuspended and cultured at low density to identify EGFP(+) progenitor cell derivatives. Under these conditions, we observed single EGFP(+) beating cells that had acquired an hyperpolarization-activated current typical of neonatal cardiomyocytes (EGFP(+) cells, -2.24 ± 0.89 pA/pF; myocytes, -1.99 ± 0.63 pA/pF, at -125 mV). To discriminate between cell autonomous differentiation and fusion, EGFP(+)/CD34(+) cells were cocultured with cardiac myocytes infected with a red fluorescence protein-lentiviral vector; under these conditions we found that 100% of EGFP(+) cells were also red fluorescent protein positive, suggesting cell fusion as the mechanism by which cardiac functional features are acquired.


Assuntos
Antígenos CD34/metabolismo , Comunicação Celular/fisiologia , Fusão Celular/métodos , Sangue Fetal/citologia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Células-Tronco/imunologia , Animais , Antígenos CD34/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Modelos Animais , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Ratos , Células-Tronco/fisiologia
12.
Cardiovasc Res ; 89(3): 650-60, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20833652

RESUMO

AIMS: Bone marrow mesenchymal stromal cell (BMStC) transplantation into the infarcted heart improves left ventricular function and cardiac remodelling. However, it has been suggested that tissue-specific cells may be better for cardiac repair than cells from other sources. The objective of the present work has been the comparison of in vitro and in vivo properties of adult human cardiac stromal cells (CStC) to those of syngeneic BMStC. METHODS AND RESULTS: Although CStC and BMStC exhibited a similar immunophenotype, their gene, microRNA, and protein expression profiles were remarkably different. Biologically, CStC, compared with BMStC, were less competent in acquiring the adipogenic and osteogenic phenotype but more efficiently expressed cardiovascular markers. When injected into the heart, in rat a model of chronic myocardial infarction, CStC persisted longer within the tissue, migrated into the scar, and differentiated into adult cardiomyocytes better than BMStC. CONCLUSION: Our findings demonstrate that although CStC and BMStC share a common stromal phenotype, CStC present cardiovascular-associated features and may represent an important cell source for more efficient cardiac repair.


Assuntos
Transplante de Medula Óssea/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Células Estromais/citologia , Adulto , Animais , Biomarcadores , Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Fusão Celular , Linhagem da Célula/fisiologia , Humanos , Imunofenotipagem , Masculino , Infarto do Miocárdio/patologia , Neovascularização Fisiológica/fisiologia , Ratos , Ratos Wistar
13.
Vet J ; 189(3): 361-3, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20822938

RESUMO

Formation of multinucleated giant cells (MGCs) by macrophage fusion is a typical cytopathic effect of lentiviral replication in caprine monocytes and MGC formation from cultured caprine peripheral blood mononuclear cells (PBMCs) has been considered to be diagnostic for small ruminant lentivirus (SRLV) infection. In this study, formation of MGCs was observed after 7-14 days when PBMCs were cultured from healthy goats free from SRLV infection. These MGCs expressed tartrate-resistant acid phosphatase, calcitonin receptor, integrin αVß3, cathepsin K and matrix metalloproteinase 9 and were able to resorb bone in vitro in the absence of RANKL and macrophage colony stimulating factor, consistent with an osteoclast phenotype.


Assuntos
Células Gigantes/patologia , Leucócitos Mononucleares/metabolismo , Monócitos/metabolismo , Osteoclastos/citologia , Fosfatase Ácida/genética , Fosfatase Ácida/metabolismo , Animais , Osso e Ossos/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Cabras , Isoenzimas/genética , Isoenzimas/metabolismo , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Fenótipo , Ligante RANK/genética , Ligante RANK/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Fosfatase Ácida Resistente a Tartarato
14.
Circ Res ; 106(3): 434-46, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20167941

RESUMO

Abstract: Pacemaking is a basic physiological process, and the cellular mechanisms involved in this function have always attracted the keen attention of investigators. The "funny" (I(f)) current, originally described in sinoatrial node myocytes as an inward current activated on hyperpolarization to the diastolic range of voltages, has properties suitable for generating repetitive activity and for modulating spontaneous rate. The degree of activation of the funny current determines, at the end of an action potential, the steepness of phase 4 depolarization; hence, the frequency of action potential firing. Because I(f) is controlled by intracellular cAMP and is thus activated and inhibited by beta-adrenergic and muscarinic M2 receptor stimulation, respectively, it represents a basic physiological mechanism mediating autonomic regulation of heart rate. Given the complexity of the cellular processes involved in rhythmic activity, an exact quantification of the extent to which I(f) and other mechanisms contribute to pacemaking is still a debated issue; nonetheless, a wealth of information collected since the current was first described more than 30 years ago clearly agrees to identify I(f) as a major player in both generation of spontaneous activity and rate control. I(f)- dependent pacemaking has recently advanced from a basic, physiologically relevant concept, as originally described, to a practical concept that has several potentially useful clinical applications and can be valuable in therapeutically relevant conditions. Typically, given their exclusive role in pacemaking, f-channels are ideal targets of drugs aiming to pharmacological control of cardiac rate. Molecules able to bind specifically to and block f-channels can thus be used as pharmacological tools for heart rate reduction with little or no adverse cardiovascular side effects. Indeed a selective f-channel inhibitor, ivabradine, is today commercially available as a tool in the treatment of stable chronic angina. Also, several loss-of-function mutations of HCN4 (hyperpolarization-activated, cyclic-nucleotide gated 4), the major constitutive subunit of f-channels in pacemaker cells, are known today to cause rhythm disturbances, such as for example inherited sinus bradycardia. Finally, gene- or cell-based methods for in situ delivery of f-channels to silent or defective cardiac muscle represent novel approaches for the development of biological pacemakers eventually able to replace electronic devices.


Assuntos
Potenciais de Ação/fisiologia , Sistema de Condução Cardíaco/fisiologia , Canais de Potássio/fisiologia , Nó Sinoatrial/fisiologia , Animais , Benzazepinas/farmacologia , AMP Cíclico/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Frequência Cardíaca/fisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/fisiologia , Ivabradina , Proteínas Musculares/fisiologia , Fosforilação , Potássio/fisiologia , Canais de Potássio/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Receptor Muscarínico M2/fisiologia , Receptores Adrenérgicos beta/fisiologia , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Nó Sinoatrial/citologia , Sódio/fisiologia
15.
Drugs ; 67 Suppl 2: 15-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17999560

RESUMO

The 'funny' (pacemaker, I(f)) current, first described almost 30 years ago in sinoatrial node (SAN) myocytes, is a mixed sodium/potassium inward current, activated on hyperpolarisation in the diastolic range of voltages. 'Funny' (f) channels are activated by intracellular cyclic adenosine monophosphate (cAMP) concentrations according to a mechanism mediating regulation of heart rate by the autonomic nervous system, as well as by voltage hyperpolarisation. Structural subunits of native f-channels are the hyperpolarisation-activated cyclic nucleotide-gated (HCN) channels; of the four HCN isoforms known, HCN4 is the most highly expressed in SAN tissue. The I(f) current is a natural target in the search for drugs aimed specifically at affecting heart rate, given its function in pacemaking. Increased heart rate has a negative influence on clinical outcome in patients with cardiovascular disease, and indeed is also an established risk factor for cardiovascular and all-cause mortality in the general population. Clearly, therefore, independent reduction of heart rate, through inhibition of the I(f) current, appears to be a suitable therapeutic option for patients with ischaemic heart disease.beta-Adrenoceptor antagonists (beta-blockers) reduce intracellular cAMP levels, and a substantial part of their negative chronotropic effect is therefore attributable to a reduction of the I(f) current. However, neither beta-blockers nor Ca(2+) channel antagonists, both of which have traditionally been used to reduce myocardial ischaemia, are 'pure' heart rate-lowering drugs. These agents may, in fact, have adverse cardiovascular and noncardiovascular effects.Conversely, the novel heart rate-reducing agent ivabradine is a specific blocker of f-channels, hence a selective inhibitor of the pacemaker I(f) current in the SAN. Ivabradine slows heart rate by reducing the I(f) current-regulated steepness of the diastolic depolarisation in SAN myocytes, thereby increasing diastolic duration, without altering action potential duration or causing negative inotropy. As such, ivabradine is particularly useful in patients with chronic stable angina pectoris. Further clinical studies are ongoing to evaluate the efficacy of ivabradine in patients with coronary heart disease, left ventricular dysfunction and heart failure. This short article reviews the current state of knowledge of the properties of the 'funny' current in relation to exploitation of the I(f) function in pacemaking generation and modulation for the pharmacological control of heart rate.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Frequência Cardíaca/fisiologia , Proteínas Musculares/fisiologia , Nó Sinoatrial/fisiologia , Animais , Benzazepinas/farmacologia , Benzazepinas/uso terapêutico , AMP Cíclico/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Eletrofisiologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Ivabradina , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Isquemia Miocárdica/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Canais de Potássio , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Nó Sinoatrial/efeitos dos fármacos
16.
Prog Biophys Mol Biol ; 90(1-3): 13-25, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-15975637

RESUMO

Since its first description in 1979 (Brown et al., 1979. Nature 280, 235-236), extensive work on the I(f) current has amply demonstrated its role in the generation and neurotransmitter-induced modulation of pacemaker activity in heart (DiFrancesco, 1993. Annual Review of Physiology 55, 455-472). At pacemaker voltages, I(f) is an inward current activated by negative voltage and by intracellular cAMP. Moderate beta-receptor stimulation accelerates, and vagal stimulation slows, cardiac rate by increasing and decreasing, respectively, I(f) at diastolic potentials via changes in cAMP level. Cloning of four isoforms of hyperpolarization-activated, cyclic-nucleotide-gated (HCN) channels in the late 1990s has shown their correlation to native f-channels. Comparison of the properties of native pacemaker channels with those of HCN channels has provided information concerning the composition and molecular features of native channels in different cardiac regions. The relevance of I(f) to pacemaker generation and modulation makes f-channels a natural target of drugs aiming to control pharmacologically heart rate. Agents selectively reducing heart rate have been developed which act by specific inhibition of I(f), such as ivabradine; these drugs have a high potential for treatment of diseases where heart rate reduction is beneficial, such as angina and heart failure. Knowledge of the molecular properties of HCN clones will help the development of drugs specifically interacting with cardiac, rather than neuronal pacemaker channels. Devices able to replace electronic pacemakers and based on the delivery of a cellular source of pacemaker channels to non-pacing tissue (biological pacemakers) are likely to be developed in the near future for use in therapies for diseases of heart rhythm.


Assuntos
Relógios Biológicos/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Coração/fisiologia , Ativação do Canal Iônico , Animais , Benzazepinas/farmacologia , Fármacos Cardiovasculares/farmacologia , AMP Cíclico/metabolismo , Humanos , Canais Iônicos/fisiologia , Ivabradina
17.
Curr Med Res Opin ; 21(7): 1115-22, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16004681

RESUMO

The 'funny' (I(f)) current, first described by Brown et al. in 1979 in pacemaker myocytes, is an inward current that slowly activates on hyperpolarization to the diastolic range of voltages. Extensive work has amply demonstrated its involvement in the generation of spontaneous activity. The extent of current activation determines the slope of diastolic depolarization and hence of pacemaker rate. Since I(f) is under cyclic adenosine monophosphate (cAMP)-mediated control by beta-adrenergic and muscarinic stimulation, this mechanism underlies neurotransmitter modulation of cardiac rate and is therefore of fundamental physiological relevance. Their key role in pacemaking makes f-channels a natural target for drugs aiming at regulation of pacemaker activity and cardiac rate. Both in the past and more recently, rate-reducing drugs that slow pacemaker activity by decreasing the rate of diastolic depolarization have been developed. These drugs act as specific f-channel inhibitors. One of the latest such molecules developed, ivabradine, has a highly specific inhibitory action on f-channels, which atypically depends on the current flow across the channel. These specific properties make the I(f) inhibition by ivabradine 'use-dependent,' a therapeutically beneficial property. Investigation of the interaction between rate-reducing molecules and specific regions of hyperpolarization-activated, cyclic nucleotidegated (HCN) channels, the molecular components of native f-channels, will provide new strategies for more specific and efficient drug design. This short review addresses the major basic properties of cardiac f-channels, with a focus on the mode of action of f-channel inhibitors and on its possible molecular interpretation.


Assuntos
Antiarrítmicos/farmacologia , Benzazepinas/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Canais Iônicos/antagonistas & inibidores , Nó Sinoatrial/efeitos dos fármacos , Sequência de Aminoácidos , Antiarrítmicos/química , Benzazepinas/química , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Desenho de Fármacos , Condutividade Elétrica , Frequência Cardíaca/fisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/química , Ivabradina , Dados de Sequência Molecular , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/química , Canais de Potássio , Nó Sinoatrial/fisiologia
18.
J Mol Cell Cardiol ; 35(8): 905-13, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12878477

RESUMO

I(f) contributes to generation and autonomic control of spontaneous activity of cardiac pacemaker cells through a cAMP-dependent, Ca(2+)-independent mechanism of rate regulation. However, disruption of Ca(2+) release from sarcoplasmic reticulum (SR) by ryanodine (Ry) has been recently shown to slow spontaneous rate and inhibit beta-adrenergic receptor (betaAR)-induced rate acceleration, leading to the suggestion that the target of betaAR modulation of pacemaking is the intracellular Ca(2+)-regulatory process. We have investigated whether the Ry-induced decrease of betaAR rate modulation alternatively involves disruption of the betaAR-adenylate-cyclase-cAMP-I(f) mechanism. Prolonged exposure to Ry (3 microM, >2 min) slowed spontaneous rate of pacemaker cells by 29.8% via a depolarizing shift of take-off potential (TOP) without significantly changing early diastolic depolarization rate. Ry depressed rate acceleration caused by isoproterenol (Iso) (1 microM, 23.6% in control vs. 8.0%), but did not modify that caused by two membrane-permeable cAMP analogs, CPT-cAMP (300 microM, 17.7% vs. 17.3%) and Rp-cAMPs (50 microM, 18.0% vs. 20.6%). Consistent with the rate effect, exposure to Ry decreased the shift induced by Iso, but not that induced by either cAMP analog on the I(f)-activation curve. We conclude that disruption of Ry receptor function and SR Ca(2+) release depresses betaAR-induced modulation of heart rate, but does not impair cAMP-dependent rate acceleration mediated by I(f). However, abolishment of normal Ca(2+) homeostasis may result in the failure of betaAR agonists to sufficiently elevate cAMP near f-channels. The molecular basis for Ca(2+)-dependent interference in beta-adrenergic signaling remains to be determined.


Assuntos
AMP Cíclico/metabolismo , Ativação do Canal Iônico , Contração Miocárdica , Rianodina/farmacologia , Nó Sinoatrial/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Cálcio/metabolismo , Diástole/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/metabolismo , Isoproterenol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Coelhos , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta/metabolismo , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Nó Sinoatrial/efeitos dos fármacos , Fatores de Tempo
19.
J Physiol ; 549(Pt 2): 347-59, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12702747

RESUMO

'Funny-' (f-) channels of cardiac sino-atrial node (SAN) cells are key players in the process of pacemaker generation and mediate the modulatory action of autonomic transmitters on heart rate. The molecular components of f-channels are the hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels. Of the four HCN isoforms known, two (HCN4 and HCN1) are expressed in the rabbit SAN at significant levels. However, the properties of f-channels of SAN cells do not conform to specific features of the two isoforms expressed locally. For example, activation kinetics and cAMP sensitivity of native pacemaker channels are intermediate between those reported for HCN1 and HCN4. Here we have explored the possibility that both HCN4 and HCN1 isoforms contribute to the native If in SAN cells by co-assembling into heteromeric channels. To this end, we used heterologous expression in human embryonic kidney (HEK) 293 cells to investigate the kinetics and cAMP response of the current generated by co-transfected (HCN4 + HCN1) and concatenated (HCN4-HCN1 (4-1) tandem or HCN1-HCN4 (1-4) tandem) rabbit constructs and compared them with those of the native f-current from rabbit SAN. 4-1 tandem, but not co-transfected, currents had activation kinetics approaching those of If; however, the activation range of 4-1 tandem channels was more negative than that of the f-channel and their cAMP sensitivity were poorer (although that of 1-4 tandem channels was normal). Co-transfection of 4-1 tandem channels with minK-related protein 1(MiRP1) did not alter their properties. HCN1 and HCN4 may contribute to native f-channels, but a 'context'-dependent mechanism is also likely to modulate the channel properties in native tissues.


Assuntos
Canais Iônicos/metabolismo , Proteínas Musculares/metabolismo , Proteínas do Tecido Nervoso , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Nó Sinoatrial/metabolismo , Animais , Linhagem Celular , AMP Cíclico/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Eletrofisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/química , Canais Iônicos/fisiologia , Cinética , Proteínas Musculares/química , Proteínas Musculares/fisiologia , Canais de Potássio/metabolismo , Coelhos , Nó Sinoatrial/citologia
20.
J Physiol ; 542(Pt 1): 89-97, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12096053

RESUMO

Gating of voltage-dependent conductances in retinal photoreceptors is the first step of a process leading to the enhancement of the temporal performance of the visual system. The molecular components underlying voltage-dependent gating in rods are presently poorly defined. In the present work we have investigated the isoform composition and the functional characteristics of hyperpolarisation-activated cyclic nucleotide-gated channels (HCN) in rabbit rods. Using immunocytochemistry we show the expression in the inner segment and cell body of the isoform 1 (HCN1). Electrophysiological investigations show that hyperpolarisation-activated currents (I(h)) can be measured only from the cell regions where HCN1 is expressed. Half-activation voltage (-75.0 +/- 0.3 mV) and kinetics (t(1/2) of 101 +/- 8 ms at -110 mV and 20 degrees C) of the I(h) in rods are similar to those of the macroscopic current carried by homomeric rabbit HCN1 channels expressed in HEK 293 cells. The homomeric nature of HCN1 channels in rods is compatible with the observation that cAMP induces a small shift (2.3 +/- 0.8 mV) in the half-activation voltage of I(h). In addition, the observation that within the physiological range of membrane potentials, cAMP does not significantly affect the gain of the current-to-voltage conversion, may reflect the need to protect the first step in the processing of visual signals from changes in cAMP turnover.


Assuntos
Canais Iônicos/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Algoritmos , Animais , Northern Blotting , Western Blotting , AMP Cíclico/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Imuno-Histoquímica , Técnicas In Vitro , Canais Iônicos/ultraestrutura , Cinética , Potenciais da Membrana/fisiologia , Microscopia Confocal , Técnicas de Patch-Clamp , Células Fotorreceptoras de Vertebrados/ultraestrutura , Canais de Potássio , RNA Mensageiro/biossíntese , Coelhos , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA